Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 52
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Arterioscler Thromb Vasc Biol ; 44(1): 290-299, 2024 01.
Artículo en Inglés | MEDLINE | ID: mdl-37970718

RESUMEN

BACKGROUND: Despite the ubiquitous utilization of central venous catheters in clinical practice, their use commonly provokes thromboembolism. No prophylactic strategy has shown sufficient efficacy to justify routine use. Coagulation factors FXI (factor XI) and FXII (factor XII) represent novel targets for device-associated thrombosis, which may mitigate bleeding risk. Our objective was to evaluate the safety and efficacy of an anti-FXI mAb (monoclonal antibody), gruticibart (AB023), in a prospective, single-arm study of patients with cancer receiving central line placement. METHODS: We enrolled ambulatory cancer patients undergoing central line placement to receive a single dose of gruticibart (2 mg/kg) administered through the venous catheter within 24 hours of placement and a follow-up surveillance ultrasound at day 14 for evaluation of catheter thrombosis. A parallel, noninterventional study was used as a comparator. RESULTS: In total, 22 subjects (n=11 per study) were enrolled. The overall incidence of catheter-associated thrombosis was 12.5% in the interventional study and 40.0% in the control study. The anti-FXI mAb, gruticibart, significantly prolonged the activated partial thromboplastin time in all subjects on day 14 compared with baseline (P<0.001). Gruticibart was well tolerated and without infusion reactions, drug-related adverse events, or clinically relevant bleeding. Platelet flow cytometry demonstrated no difference in platelet activation following administration of gruticibart. T (thrombin)-AT (antithrombin) and activated FXI-AT complexes increased following central line placement in the control study, which was not demonstrated in our intervention study. CRP (C-reactive protein) did not significantly increase on day 14 in those who received gruticibart, but it did significantly increase in the noninterventional study. CONCLUSIONS: FXI inhibition with gruticibart was well tolerated without any significant adverse or bleeding-related events and resulted in a lower incidence of catheter-associated thrombosis on surveillance ultrasound compared with the published literature and our internal control study. These findings suggest that targeting FXI could represent a safe intervention to prevent catheter thrombosis. REGISTRATION: URL: https://www.clinicaltrials.gov; Unique identifier: NCT04465760.


Asunto(s)
Neoplasias , Trombosis , Humanos , Factor XI/metabolismo , Estudios Prospectivos , Trombosis/etiología , Trombosis/prevención & control , Trombosis/tratamiento farmacológico , Hemorragia/inducido químicamente , Catéteres/efectos adversos , Neoplasias/tratamiento farmacológico , Neoplasias/complicaciones
2.
Am J Physiol Cell Physiol ; 326(1): C40-C49, 2024 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-37955120

RESUMEN

The blood-brain barrier is composed of microvascular endothelial cells, immune cells, and astrocytes that work in concert with the coagulation cascade to control inflammation and immune cell infiltration into the central nervous system. Endothelial cell dysfunction leading to increased permeability and compromised barrier function are hallmarks of neuroinflammatory and autoimmune disorders, including multiple sclerosis (MS). Therapeutic strategies that improve or protect endothelial barrier function may be beneficial in the treatment or prevention of neuroinflammatory diseases. We therefore tested the hypothesis that biasing thrombin toward anticoagulant and cytoprotective activities would provide equivalent or even additive benefit compared with standard-of-care therapeutic strategies, including corticosteroids. In a mouse model of relapsing-remitting MS, treatment with the thrombin mutant, E-WE thrombin, an engineered thrombin mutant with cytoprotective activities that is biased toward anticoagulant and cytoprotective activity, reduced neuroinflammation and extracellular fibrin formation in SJL mice inoculated with proteolipid protein (PLP) peptide. When administered at the onset of detectable disease, E-WE thrombin significantly improved the disease severity of the initial attack as well as the relapse and delayed the onset of relapse to a similar extent as observed with methylprednisolone. Both methylprednisolone and E-WE thrombin reduced demyelination and immune cell recruitment. These results provide rationale for considering engineered forms of thrombin biased toward anticoagulant and cytoprotective activity as a therapeutic strategy and perhaps an effective alternative to high-dose methylprednisolone for the management of acute relapsing MS attacks.NEW & NOTEWORTHY There are limited treatment options for mitigating acute relapsing attacks for patients with multiple sclerosis. We tested the hypothesis that harnessing the cytoprotective activity of the blood coagulation enzyme, thrombin, would provide benefit and protection against relapsing disease in a mouse model of MS. Our results provide rationale for considering engineered forms of thrombin biased toward cytoprotective activity as a therapeutic strategy and perhaps an alternative to steroids for the management of relapsing MS attacks.


Asunto(s)
Esclerosis Múltiple Recurrente-Remitente , Trombina , Animales , Humanos , Ratones , Anticoagulantes , Modelos Animales de Enfermedad , Células Endoteliales/metabolismo , Metilprednisolona , Esclerosis Múltiple Recurrente-Remitente/tratamiento farmacológico , Gravedad del Paciente , Recurrencia , Trombina/uso terapéutico
3.
Blood ; 138(22): 2173-2184, 2021 12 02.
Artículo en Inglés | MEDLINE | ID: mdl-34086880

RESUMEN

End-stage renal disease (ESRD) patients on chronic hemodialysis have repeated blood exposure to artificial surfaces that can trigger clot formation within the hemodialysis circuit. Dialyzer clotting can lead to anemia despite erythropoietin and iron supplementation. Unfractionated heparin prevents clotting during hemodialysis, but it is not tolerated by all patients. Although heparin-free dialysis is performed, intradialytic blood entrapment can be problematic. To address this issue, we performed a randomized, double-blind, phase 2 study comparing AB023, a unique antibody that binds factor XI (FXI) and blocks its activation by activated FXII, but not by thrombin, to placebo in 24 patients with ESRD undergoing heparin-free hemodialysis. Patients were randomized to receive a single predialysis dose of AB023 (0.25 or 0.5 mg/kg) or placebo in a 2:1 ratio, and safety and preliminary efficacy were compared with placebo and observations made prior to dosing within each treatment arm. AB023 administration was not associated with impaired hemostasis or other drug-related adverse events. Occlusive events requiring hemodialysis circuit exchange were less frequent and levels of thrombin-antithrombin complexes and C-reactive protein were lower after AB023 administration compared with data collected prior to dosing. AB023 also reduced potassium and iron entrapment in the dialyzers, consistent with less blood accumulation within the dialyzers. We conclude that despite the small sample size, inhibition of contact activation-induced coagulation with AB023 was well tolerated and reduced clotting within the dialyzer. This trial was registered at www.clinicaltrials.gov as #NCT03612856.


Asunto(s)
Anticuerpos Monoclonales Humanizados/uso terapéutico , Antitrombinas/uso terapéutico , Fallo Renal Crónico/terapia , Diálisis Renal/métodos , Adulto , Anticuerpos Monoclonales Humanizados/efectos adversos , Antitrombinas/efectos adversos , Método Doble Ciego , Factor XI/antagonistas & inhibidores , Femenino , Hemostasis/efectos de los fármacos , Humanos , Masculino , Persona de Mediana Edad , Efecto Placebo , Diálisis Renal/efectos adversos , Trombosis/etiología , Trombosis/prevención & control
4.
Blood ; 138(2): 178-189, 2021 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-33598692

RESUMEN

Activation of coagulation factor (F) XI promotes multiorgan failure in rodent models of sepsis and in a baboon model of lethal systemic inflammation induced by infusion of heat-inactivated Staphylococcus aureus. Here we used the anticoagulant FXII-neutralizing antibody 5C12 to verify the mechanistic role of FXII in this baboon model. Compared with untreated control animals, repeated 5C12 administration before and at 8 and 24 hours after bacterial challenge prevented the dramatic increase in circulating complexes of contact system enzymes FXIIa, FXIa, and kallikrein with antithrombin or C1 inhibitor, and prevented cleavage and consumption of high-molecular-weight kininogen. Activation of several coagulation factors and fibrinolytic enzymes was also prevented. D-dimer levels exhibited a profound increase in the untreated animals but not in the treated animals. The antibody also blocked the increase in plasma biomarkers of inflammation and cell damage, including tumor necrosis factor, interleukin (IL)-1ß, IL-6, IL-8, IL-10, granulocyte-macrophage colony-stimulating factor, nucleosomes, and myeloperoxidase. Based on clinical presentation and circulating biomarkers, inhibition of FXII prevented fever, terminal hypotension, respiratory distress, and multiorgan failure. All animals receiving 5C12 had milder and transient clinical symptoms and were asymptomatic at day 7, whereas untreated control animals suffered irreversible multiorgan failure and had to be euthanized within 2 days after the bacterial challenge. This study confirms and extends our previous finding that at least 2 enzymes of the contact activation complex, FXIa and FXIIa, play critical roles in the development of an acute and terminal inflammatory response in baboons challenged with heat-inactivated S aureus.


Asunto(s)
Factor XII/metabolismo , Insuficiencia Multiorgánica/metabolismo , Insuficiencia Multiorgánica/microbiología , Staphylococcus aureus/fisiología , Animales , Anticuerpos/uso terapéutico , Trastornos de la Coagulación Sanguínea/complicaciones , Trastornos de la Coagulación Sanguínea/inmunología , Trastornos de la Coagulación Sanguínea/microbiología , Plaquetas/metabolismo , Microambiente Celular , Activación de Complemento , Factor XII/inmunología , Femenino , Fibrinógeno/metabolismo , Calor , Inflamación/complicaciones , Inflamación/patología , Masculino , Insuficiencia Multiorgánica/inmunología , Papio , Infecciones Estafilocócicas/tratamiento farmacológico , Infecciones Estafilocócicas/inmunología , Infecciones Estafilocócicas/microbiología , Infecciones Estafilocócicas/patología , Análisis de Supervivencia
5.
J Immunol ; 206(8): 1784-1792, 2021 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-33811105

RESUMEN

Complement factor H (CFH) is the major inhibitor of the alternative pathway of the complement system and is structurally related to beta2-glycoprotein I, which itself is known to bind to ligands, including coagulation factor XI (FXI). We observed reduced complement activation when FXI activation was inhibited in a baboon model of lethal systemic inflammation, suggesting cross-talk between FXI and the complement cascade. It is unknown whether FXI or its activated form, activated FXI (FXIa), directly interacts with the complement system. We explored whether FXI could interact with and inhibit the activity of CFH. We found that FXIa neutralized CFH by cleavage of the R341/R342 bonds. FXIa reduced the capacity of CFH to enhance the cleavage of C3b by factor I and the decay of C3bBb. The binding of CFH to human endothelial cells was also reduced after incubating CFH with FXIa. The addition of either short- or long-chain polyphosphate enhanced the capacity of FXIa to cleave CFH. FXIa also cleaved CFH that was present on endothelial cells and in the secretome from blood platelets. The generation of FXIa in plasma induced the cleavage of CFH. Moreover, FXIa reduced the cleavage of C3b by factor I in serum. Conversely, we observed that CFH inhibited FXI activation by either thrombin or FXIIa. Our study provides, to our knowledge, a novel molecular link between the contact pathway of coagulation and the complement system. These results suggest that FXIa generation enhances the activity of the complement system and thus may potentiate the immune response.


Asunto(s)
Plaquetas/metabolismo , Factor H de Complemento/metabolismo , Células Endoteliales/metabolismo , Factor XIa/metabolismo , Inflamación/metabolismo , Animales , Coagulación Sanguínea , Complemento C3b/metabolismo , Vía Alternativa del Complemento , Fibrinógeno/metabolismo , Células Endoteliales de la Vena Umbilical Humana , Humanos , Papio , Unión Proteica , Receptor Cross-Talk
6.
Metab Brain Dis ; 38(7): 2383-2391, 2023 10.
Artículo en Inglés | MEDLINE | ID: mdl-37341855

RESUMEN

Multiple sclerosis (MS) is the most common causes of non-traumatic disability in young adults worldwide. MS pathophysiologies include the formation of inflammatory lesions, axonal damage and demyelination, and blood brain barrier (BBB) disruption. Coagulation proteins, including factor (F)XII, can serve as important mediators of the adaptive immune response during neuroinflammation. Indeed, plasma FXII levels are increased during relapse in relapsing-remitting MS patients, and previous studies showed that reducing FXII levels was protective in a murine model of MS, experimental autoimmune encephalomyelitis (EAE). Our objective was to determine if pharmacological targeting of FXI, a major substrate of activated FXII (FXIIa), improves neurological function and attenuates CNS damage in the setting of EAE. EAE was induced in male mice using murine myelin oligodendrocyte glycoprotein peptides combined with heat-inactivated Mycobacterium tuberculosis and pertussis toxin. Upon onset of symptoms, mice were treated every other day intravenously with anti-FXI antibody, 14E11, or saline. Disease scores were recorded daily until euthanasia for ex vivo analyses of inflammation. Compared to the vehicle control, 14E11 treatment reduced the clinical severity of EAE and total mononuclear cells, including CD11b+CD45high macrophage/microglia and CD4+ T cell numbers in brain. Following pharmacological targeting of FXI, BBB disruption was reduced, as measured by decreased axonal damage and fibrin(ogen) accumulation in the spinal cord. These data demonstrate that pharmacological inhibition of FXI reduces disease severity, immune cell migration, axonal damage, and BBB disruption in mice with EAE. Thus, therapeutic agents targeting FXI and FXII may provide a useful approach for treating autoimmune and neurologic disorders.


Asunto(s)
Encefalomielitis Autoinmune Experimental , Esclerosis Múltiple , Animales , Masculino , Ratones , Encéfalo/metabolismo , Encefalomielitis Autoinmune Experimental/metabolismo , Factor XI/antagonistas & inhibidores , Factor XI/metabolismo , Ratones Endogámicos C57BL , Esclerosis Múltiple/patología , Glicoproteína Mielina-Oligodendrócito , Médula Espinal/metabolismo
7.
Blood ; 135(9): 689-699, 2020 02 27.
Artículo en Inglés | MEDLINE | ID: mdl-31977000

RESUMEN

Although thrombin is a key enzyme in the coagulation cascade and is required for both normal hemostasis and pathologic thrombogenesis, it also participates in its own negative feedback via activation of protein C, which downregulates thrombin generation by enzymatically inactivating factors Va and VIIIa. Our group and others have previously shown that thrombin's procoagulant and anticoagulant activities can be effectively disassociated to varying extents through site-directed mutagenesis. The thrombin mutant W215A/E217A (WE thrombin) has been one of the best characterized constructs with selective activity toward protein C. Although animal studies have demonstrated that WE thrombin acts as an anticoagulant through activated protein C (APC) generation, the observed limited systemic anticoagulation does not fully explain the antithrombotic potency of this or other thrombin mutants. AB002 (E-WE thrombin) is an investigational protein C activator thrombin analog in phase 2 clinical development (clinicaltrials.gov NCT03963895). Here, we demonstrate that this molecule is a potent enzyme that is able to rapidly interrupt arterial-type thrombus propagation at exceedingly low doses (<2 µg/kg, IV), yet without substantial systemic anticoagulation in baboons. We demonstrate that AB002 produces APC on platelet aggregates and competitively inhibits thrombin-activatable fibrinolysis inhibitor (carboxypeptidase B2) activation in vitro, which may contribute to the observed in vivo efficacy. We also describe its safety and activity in a phase 1 first-in-human clinical trial. Together, these results support further clinical evaluation of AB002 as a potentially safe and effective new approach for treating or preventing acute thrombotic and thromboembolic conditions. This trial was registered at www.clinicaltrials.gov as #NCT03453060.


Asunto(s)
Fibrinolíticos/farmacología , Proteína C/efectos de los fármacos , Trombina/análogos & derivados , Trombosis/prevención & control , Adulto , Animales , Método Doble Ciego , Humanos , Persona de Mediana Edad , Papio , Proteínas Recombinantes/farmacología
8.
Am J Physiol Cell Physiol ; 320(3): C365-C374, 2021 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-33471623

RESUMEN

Factor XI (FXI) has been shown to bind platelets, but the functional significance of this observation remains unknown. Platelets are essential for hemostasis and play a critical role in thrombosis, whereas FXI is not essential for hemostasis but promotes thrombosis. An apparent functional contradiction, platelets are known to support thrombin generation, yet platelet granules release protease inhibitors, including those of activated FXI (FXIa). We aim to investigate the secretory and binding mechanisms by which platelets could support or inhibit FXIa activity. The presence of platelets enhanced FXIa activity in a purified system and increased coagulation Factor IX (FIX) activation by FXIa and fibrin generation in human plasma. In contrast, platelets reduced the activation of FXI by activated coagulation factor XII (FXIIa) and the activation of FXII by kallikrein (PKa). Incubation of FXIa with the platelet secretome, which contains FXIa inhibitors, such as protease nexin-II, abolished FXIa activity, yet in the presence of activated platelets, the secretome was not able to block the activity of FXIa. FXIa variants lacking the anion-binding sites did not alter the effect of platelets on FXIa activity or interaction. Western blot analysis of bound FXIa [by FXIa-platelet membrane immunoprecipitation] showed that the interaction with platelets is zinc dependent and, unlike FXI binding to platelets, not dependent on glycoprotein Ib. FXIa binding to the platelet membrane increases its capacity to activate FIX in plasma likely by protecting it from inhibition by inhibitors secreted by activated platelets. Our findings suggest that an interaction of FXIa with the platelet surface may induce an allosteric modulation of FXIa.


Asunto(s)
Plaquetas/metabolismo , Factor XIa/metabolismo , Adolescente , Precursor de Proteína beta-Amiloide/metabolismo , Sitios de Unión/fisiología , Coagulación Sanguínea/fisiología , Femenino , Hemostasis/fisiología , Humanos , Masculino , Trombina/metabolismo , Trombosis/metabolismo
9.
Arterioscler Thromb Vasc Biol ; 39(4): 799-809, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30700130

RESUMEN

Objective- Factor XI (FXI) contributes to thrombotic disease while playing a limited role in normal hemostasis. We generated a unique, humanized anti-FXI antibody, AB023, which blocks factor XIIa-mediated FXI activation without inhibiting FXI activation by thrombin or the procoagulant function of FXIa. We sought to confirm the antithrombotic activity of AB023 in a baboon thrombosis model and to evaluate the safety, tolerability, pharmacokinetics, and pharmacodynamics in healthy adult subjects. Approach and Results- In a primate model of acute vascular graft thrombosis, AB023 reduced platelet and fibrin accumulation within the grafts by >75%. To evaluate the safety of AB023, we performed a first-in-human study in healthy adult volunteers without any serious adverse events. Overall, 10 of 21 (48%) subjects experienced 20 treatment-emergent adverse events, with 7 of 16 (44%) subjects following active treatment and 3 of 5 (60%) subjects following placebo. AB023 did not increase bleeding or prothrombin times. Anticoagulation was verified by a saturable ≈2-fold prolongation of the partial thromboplastin time for over 1 month after the highest dose. Conclusions- AB023, which inhibits contact activation-initiated blood coagulation in vitro and experimental thrombus formation in primates, produced a dose-dependent duration of limited anticoagulation without drug-related adverse effects in a phase 1 trial. When put in context with earlier observations suggesting that FXI contributes to venous thromboembolism and cardiovascular disease, although contributing minimally to hemostasis, our data further justify clinical evaluation of AB023 in conditions where contact-initiated FXI activation is suspected to have a pathogenic role. Clinical Trial Registration- URL: http://www.clinicaltrials.gov . Unique identifier: NCT03097341.


Asunto(s)
Anticuerpos Monoclonales Humanizados/uso terapéutico , Anticoagulantes/uso terapéutico , Factor XI/antagonistas & inhibidores , Factor XIa/fisiología , Fibrinolíticos/uso terapéutico , Adulto , Animales , Anticuerpos Monoclonales Humanizados/efectos adversos , Anticuerpos Monoclonales Humanizados/inmunología , Anticuerpos Monoclonales Humanizados/farmacología , Anticoagulantes/efectos adversos , Anticoagulantes/inmunología , Anticoagulantes/farmacología , Área Bajo la Curva , Relación Dosis-Respuesta a Droga , Método Doble Ciego , Factor XI/inmunología , Factor XIIa/fisiología , Fibrinolíticos/efectos adversos , Fibrinolíticos/inmunología , Fibrinolíticos/farmacología , Oclusión de Injerto Vascular/tratamiento farmacológico , Humanos , Papio , Tiempo de Tromboplastina Parcial
10.
Arterioscler Thromb Vasc Biol ; 38(8): 1748-1760, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-30354195

RESUMEN

Objective- Terminal complications of bacterial sepsis include development of disseminated intravascular consumptive coagulopathy. Bacterial constituents, including long-chain polyphosphates (polyP), have been shown to activate the contact pathway of coagulation in plasma. Recent work shows that activation of the contact pathway in flowing whole blood promotes thrombin generation and platelet activation and consumption distal to thrombus formation ex vivo and in vivo. Here, we sought to determine whether presence of long-chain polyP or bacteria in the bloodstream promotes platelet activation and consumption in a coagulation factor (F)XII-dependent manner. Approach and Results- Long-chain polyP promoted platelet P-selectin expression, microaggregate formation, and platelet consumption in flowing whole blood in a contact activation pathway-dependent manner. Moreover, long-chain polyP promoted local fibrin formation on collagen under shear flow in a FXI-dependent manner. Distal to the site of thrombus formation, platelet consumption was dramatically enhanced in the presence of long-chain polyP in the blood flow in a FXI- and FXII-dependent manner. In a murine model, long-chain polyP promoted platelet deposition and fibrin generation in lungs in a FXII-dependent manner. In a nonhuman primate model of bacterial sepsis, pre-treatment of animals with an antibody blocking FXI activation by FXIIa reduced lethal dose100 Staphylococcus aureus-induced platelet and fibrinogen consumption. Conclusions- This study demonstrates that bacterial-type long-chain polyP promotes platelet activation in a FXII-dependent manner in flowing blood, which may contribute to sepsis-associated thrombotic processes, consumptive coagulopathy, and thrombocytopenia.


Asunto(s)
Coagulación Sanguínea/efectos de los fármacos , Plaquetas/efectos de los fármacos , Factor XII/metabolismo , Factor XIIa/metabolismo , Activación Plaquetaria/efectos de los fármacos , Polifosfatos/toxicidad , Trombosis/inducido químicamente , Animales , Plaquetas/metabolismo , Modelos Animales de Enfermedad , Factor XII/genética , Factor XIIa/genética , Femenino , Fibrina/metabolismo , Humanos , Masculino , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Papio ursinus , Precalicreína/genética , Precalicreína/metabolismo , Embolia Pulmonar/sangre , Embolia Pulmonar/inducido químicamente , Embolia Pulmonar/genética , Sepsis/sangre , Sepsis/microbiología , Transducción de Señal/efectos de los fármacos , Infecciones Estafilocócicas/sangre , Infecciones Estafilocócicas/microbiología , Trombosis/sangre , Trombosis/genética , Calicreínas de Tejido/genética , Calicreínas de Tejido/metabolismo
11.
Blood ; 136(14): 1576-1577, 2020 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-33002124
12.
Blood ; 125(9): 1488-96, 2015 Feb 26.
Artículo en Inglés | MEDLINE | ID: mdl-25587039

RESUMEN

Activation of coagulation factor XI (FXI) may play a role in hemostasis. The primary substrate of activated FXI (FXIa) is FIX, leading to FX activation (FXa) and thrombin generation. However, recent studies suggest the hemostatic role of FXI may not be restricted to the activation of FIX. We explored whether FXI could interact with and inhibit the activity of tissue factor pathway inhibitor (TFPI). TFPI is an essential reversible inhibitor of activated factor X (FXa) and also inhibits the FVIIa-TF complex. We found that FXIa neutralized both endothelium- and platelet-derived TFPI by cleaving the protein between the Kunitz (K) 1 and K2 domains (Lys86/Thr87) and at the active sites of the K2 (Arg107/Gly108) and K3 (Arg199/Ala200) domains. Addition of FXIa to plasma was able to reverse the ability of TFPI to prolong TF-initiated clotting times in FXI- or FIX-deficient plasma, as well as FXa-initiated clotting times in FX-deficient plasma. Treatment of cultured endothelial cells with FXIa increased the generation of FXa and promoted TF-dependent fibrin formation in recalcified plasma. Together, these results suggest that the hemostatic role of FXIa may be attributed not only to activation of FIX but also to promoting the extrinsic pathway of thrombin generation through inactivation of TFPI.


Asunto(s)
Coagulación Sanguínea/fisiología , Plaquetas/metabolismo , Factor IX/metabolismo , Factor XIa/metabolismo , Factor Xa/metabolismo , Fibrina/metabolismo , Lipoproteínas/metabolismo , Plaquetas/citología , Western Blotting , Células Cultivadas , Citometría de Flujo , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Lipoproteínas/genética , Mutación/genética , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo
13.
Arterioscler Thromb Vasc Biol ; 36(3): 510-7, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26769048

RESUMEN

OBJECTIVE: Coagulation factor XI (FXI) has been shown to contribute to thrombus formation on collagen or tissue factor-coated surfaces in vitro and in vivo by enhancing thrombin generation. Whether the role of the intrinsic pathway of coagulation is restricted to the local site of thrombus formation is unknown. This study was aimed to determine whether FXI could promote both proximal and distal platelet activation and aggregate formation in the bloodstream. APPROACH AND RESULTS: Pharmacological blockade of FXI activation or thrombin activity in blood did not affect local platelet adhesion, yet reduced local platelet aggregation, thrombin localization, and fibrin formation on immobilized collagen and tissue factor under shear flow, ex vivo. Downstream of the thrombus formed on immobilized collagen or collagen and 10 pmol/L tissue factor, platelet CD62P expression, microaggregate formation, and progressive platelet consumption were significantly reduced in the presence of FXI function-blocking antibodies or a thrombin inhibitor in a shear rate- and time-dependent manner. In a non-human primate model of thrombus formation, we found that inhibition of FXI reduced single platelet consumption in the bloodstream distal to a site of thrombus formation. CONCLUSIONS: This study demonstrates that the FXI-thrombin axis contributes to distal platelet activation and procoagulant microaggregate formation in the blood flow downstream of the site of thrombus formation. Our data highlight FXI as a novel therapeutic target for inhibiting distal platelet consumption without affecting proximal platelet adhesion.


Asunto(s)
Coagulación Sanguínea , Plaquetas/metabolismo , Factor XI/metabolismo , Mecanotransducción Celular , Activación Plaquetaria , Trombosis/sangre , Animales , Colágeno/sangre , Modelos Animales de Enfermedad , Factor XIa/metabolismo , Fibrina/metabolismo , Humanos , Masculino , Selectina-P/sangre , Papio anubis , Agregación Plaquetaria , Flujo Sanguíneo Regional , Estrés Mecánico , Trombina/metabolismo , Tromboplastina/metabolismo , Trombosis/fisiopatología , Trombosis/prevención & control , Factores de Tiempo
14.
Blood ; 123(11): 1739-46, 2014 Mar 13.
Artículo en Inglés | MEDLINE | ID: mdl-24408325

RESUMEN

The plasma zymogens factor XII (fXII) and factor XI (fXI) contribute to thrombosis in a variety of mouse models. These proteins serve a limited role in hemostasis, suggesting that antithrombotic therapies targeting them may be associated with low bleeding risks. Although there is substantial epidemiologic evidence supporting a role for fXI in human thrombosis, the situation is not as clear for fXII. We generated monoclonal antibodies (9A2 and 15H8) against the human fXII heavy chain that interfere with fXII conversion to the protease factor XIIa (fXIIa). The anti-fXII antibodies were tested in models in which anti-fXI antibodies are known to have antithrombotic effects. Both anti-fXII antibodies reduced fibrin formation in human blood perfused through collagen-coated tubes. fXII-deficient mice are resistant to ferric chloride-induced arterial thrombosis, and this resistance can be reversed by infusion of human fXII. 9A2 partially blocks, and 15H8 completely blocks, the prothrombotic effect of fXII in this model. 15H8 prolonged the activated partial thromboplastin time of baboon and human plasmas. 15H8 reduced fibrin formation in collagen-coated vascular grafts inserted into arteriovenous shunts in baboons, and reduced fibrin and platelet accumulation downstream of the graft. These findings support a role for fXII in thrombus formation in primates.


Asunto(s)
Modelos Animales de Enfermedad , Deficiencia del Factor XII/complicaciones , Factor XII/antagonistas & inhibidores , Factor XII/fisiología , Trombina/metabolismo , Trombosis/prevención & control , Animales , Anticuerpos Monoclonales/inmunología , Anticuerpos Monoclonales/metabolismo , Coagulación Sanguínea , Factor XI/metabolismo , Factor XIIa/metabolismo , Fibrina/metabolismo , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Papio , Proteínas Recombinantes/genética , Proteínas Recombinantes/inmunología , Proteínas Recombinantes/metabolismo , Tromboplastina/metabolismo , Trombosis/etiología , Trombosis/metabolismo
15.
Metab Brain Dis ; 30(1): 57-65, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24810631

RESUMEN

Multiple sclerosis (MS) is a neuroinflammatory disease characterized by demyelination and axonal damage of the central nervous system. The pathogenesis of MS has also been linked to vascular inflammation and local activation of the coagulation system, resulting in perivascular fibrin deposition. Treatment of experimental autoimmune encephalomyelitis (EAE), a model of human MS, with antithrombotic and antiinflammatory activated protein C (APC) reduces disease severity. Since recombinant APC (Drotecogin alfa), originally approved for the treatment of severe sepsis, is not available for human MS studies, we tested the hypothesis that pharmacologic activation of endogenous protein C could likewise improve the outcome of EAE. Mice were immunized with murine myelin oligodendrocyte glycoprotein (MOG) peptides and at the onset of EAE symptoms, were treated every other day with either WE thrombin (25 µg/kg; i.v.), a selective recombinant protein C activator thrombin analog, or saline control. Mice were monitored for changes in disease score until euthanized for ex vivo analysis of inflammation. Administration of WE thrombin significantly ameliorated clinical severity of EAE, reduced inflammatory cell infiltration and demyelination, suppressed the activation of macrophages comprising the CD11b + population and reduced accumulation of fibrin (ogen) in the spinal cord. These data suggest that symptomatic MS may respond to a treatment strategy that involves temporal pharmacological enhancement of endogenous APC generation.


Asunto(s)
Encefalomielitis Autoinmune Experimental/tratamiento farmacológico , Proteína C/agonistas , Trombina/uso terapéutico , Animales , Evaluación Preclínica de Medicamentos , Encefalomielitis Autoinmune Experimental/etiología , Encefalomielitis Autoinmune Experimental/patología , Activación Enzimática , Fibrina/análisis , Fibrinógeno/análisis , Humanos , Molécula 1 de Adhesión Intercelular/biosíntesis , Activación de Macrófagos , Masculino , Ratones , Esclerosis Múltiple , Glicoproteína Mielina-Oligodendrócito/inmunología , Fragmentos de Péptidos/inmunología , Mutación Puntual , Proteína C/metabolismo , Médula Espinal/patología , Bazo/inmunología , Bazo/patología , Trombina/genética , Resultado del Tratamiento , Factor de Necrosis Tumoral alfa/biosíntesis , Sustancia Blanca/patología
16.
Blood ; 119(20): 4762-8, 2012 May 17.
Artículo en Inglés | MEDLINE | ID: mdl-22442348

RESUMEN

Severe bacterial sepsis often leads to a systemic procoagulant and proinflammatory condition that can manifest as disseminated intravascular coagulation, septic shock, and multiple organ failure. Because activation of the contact proteases factor XII (FXII), prekallikrein, and factor XI (FXI) can trigger coagulation and inflammatory responses, the contact factors have been considered potential targets for the treatment of sepsis. However, the pathogenic role of contact activation in severe infections has not been well defined. We therefore investigated whether an anticoagulant antibody (14E11) that selectively inhibits prothrombotic FXI activation by activated FXII (FXIIa) modifies the course of bowel perforation-induced peritoneal sepsis in mice. Early anticoagulation with 14E11 suppressed systemic thrombin- antithrombin complex formation, IL-6, and TNF-α levels, and reduced platelet consumption in the circulation and deposition in the blood vessels. Treatment with 14E11 within 12 hours after bowel perforation significantly improved survival compared with vehicle treatment, and the saturating dose did not increase tail bleeding. These data suggest that severe polymicrobial abdominal infection induces prothrombotic FXI activation, to the detriment of the host. Systemic anticoagulation by inhibiting FXI activation or FXIIa procoagulant activity during sepsis may therefore limit the development of disseminated intravascular coagulation without increasing bleeding risks.


Asunto(s)
Anticoagulantes/farmacología , Trastornos de la Coagulación Sanguínea/prevención & control , Coinfección/tratamiento farmacológico , Coinfección/mortalidad , Inflamación/prevención & control , Sepsis/tratamiento farmacológico , Sepsis/mortalidad , Animales , Anticuerpos Monoclonales/farmacología , Anticuerpos Monoclonales/uso terapéutico , Anticoagulantes/uso terapéutico , Trastornos de la Coagulación Sanguínea/tratamiento farmacológico , Trastornos de la Coagulación Sanguínea/etiología , Trastornos de la Coagulación Sanguínea/mortalidad , Coinfección/complicaciones , Coinfección/patología , Regulación hacia Abajo/efectos de los fármacos , Factor XIa/antagonistas & inhibidores , Factor XIa/inmunología , Inmunoterapia , Inflamación/etiología , Inflamación/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Proteína C/farmacología , Proteína C/uso terapéutico , Sepsis/complicaciones , Sepsis/patología , Análisis de Supervivencia
17.
Arterioscler Thromb Vasc Biol ; 33(7): 1670-8, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23559626

RESUMEN

OBJECTIVE: During coagulation, factor IX (FIX) is activated by 2 distinct mechanisms mediated by the active proteases of either FVIIa or FXIa. Both coagulation factors may contribute to thrombosis; FXI, however, plays only a limited role in the arrest of bleeding. Therefore, therapeutic targeting of FXI may produce an antithrombotic effect with relatively low hemostatic risk. APPROACH AND RESULTS: We have reported that reducing FXI levels with FXI antisense oligonucleotides produces antithrombotic activity in mice, and that administration of FXI antisense oligonucleotides to primates decreases circulating FXI levels and activity in a dose-dependent and time-dependent manner. Here, we evaluated the relationship between FXI plasma levels and thrombogenicity in an established baboon model of thrombosis and hemostasis. In previous studies with this model, antibody-induced inhibition of FXI produced potent antithrombotic effects. In the present article, antisense oligonucleotides-mediated reduction of FXI plasma levels by ≥ 50% resulted in a demonstrable and sustained antithrombotic effect without an increased risk of bleeding. CONCLUSIONS: These results indicate that reducing FXI levels using antisense oligonucleotides is a promising alternative to direct FXI inhibition, and that targeting FXI may be potentially safer than conventional antithrombotic therapies that can markedly impair primary hemostasis.


Asunto(s)
Coagulación Sanguínea/efectos de los fármacos , Factor XI/metabolismo , Fibrinolíticos/administración & dosificación , Oligonucleótidos Antisentido/administración & dosificación , Trombosis/prevención & control , Animales , Anticuerpos Monoclonales/administración & dosificación , Derivación Arteriovenosa Quirúrgica , Tiempo de Sangría , Colágeno , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Regulación hacia Abajo , Factor XI/antagonistas & inhibidores , Factor XI/genética , Fibrinolíticos/toxicidad , Hemorragia/inducido químicamente , Macaca fascicularis , Oligonucleótidos Antisentido/toxicidad , Papio , Trombina/metabolismo , Trombosis/sangre , Trombosis/etiología , Trombosis/genética , Factores de Tiempo
18.
J Thromb Haemost ; 22(5): 1433-1446, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38331196

RESUMEN

BACKGROUND: Cardiovascular implantable devices, such as vascular stents, are critical for the treatment of cardiovascular diseases. However, their success is dependent on robust and often long-term antithrombotic therapies. Yet, the current standard-of-care therapies often pose significant bleeding risks to patients. Coagulation factor (F)XI and FXII have emerged as potentially safe and efficacious targets to safely reduce pathologic thrombin generation in medical devices. OBJECTIVES: To study the efficacy of monoclonal antibody-targeting FXII and FXI of the contact pathway in preventing vascular device-related thrombosis. METHODS: The effects of inhibition of FXII and FXI using function-blocking monoclonal antibodies were examined in a nonhuman primate model of nitinol stent-related thrombosis under arterial and venous flow conditions. RESULTS: We found that function-blocking antibodies of FXII and FXI reduced markers of stent-induced thrombosis in vitro and ex vivo. However, FXI inhibition resulted in more effective mitigation of thrombosis markers under varied flow conditions. CONCLUSION: This work provides further support for the translation of contact pathway of coagulation inhibitors for their adjunctive clinical use with cardiovascular devices.


Asunto(s)
Aleaciones , Anticuerpos Monoclonales , Factor XII , Factor XI , Stents , Trombosis , Animales , Trombosis/prevención & control , Trombosis/sangre , Factor XII/metabolismo , Factor XII/antagonistas & inhibidores , Factor XII/inmunología , Factor XI/antagonistas & inhibidores , Factor XI/inmunología , Factor XI/metabolismo , Anticuerpos Monoclonales/farmacología , Humanos , Coagulación Sanguínea/efectos de los fármacos , Modelos Animales de Enfermedad , Masculino , Flujo Sanguíneo Regional , Fibrinolíticos/farmacología
19.
Res Pract Thromb Haemost ; 8(1): 102276, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-38226339

RESUMEN

Background: Hyperlipidemia is associated with chronic inflammation and thromboinflammation. This is an underlying cause of several cardiovascular diseases, including atherosclerosis. In diseased blood vessels, rampant thrombin generation results in the initiation of the coagulation cascade, activation of platelets, and endothelial cell dysfunction. Coagulation factor (F) XI represents a promising therapeutic target to reduce thromboinflammation, as it is uniquely positioned at an intersection between inflammation and thrombin generation. Objectives: This study aimed to investigate the role of FXI in promoting platelet and endothelial cell activation in a model of hyperlipidemia. Methods: Nonhuman primates (NHPs) were fed a standard chow diet (lean, n = 6) or a high-fat diet (obese, n = 8) to establish a model of hyperlipidemia. Obese NHPs were intravenously administered a FXI blocking antibody (2 mg/kg) and studied at baseline and at 1, 7, 14, 21, and 28 days after drug administration. Platelet activation and inflammatory markers were measured using fluorescence-activated cell sorting or enzyme-linked immunosorbent assay. Molecular imaging was used to quantify vascular cell adhesion molecule 1 (VCAM-1) expression at the carotid bifurcation. Results: Obese NHPs demonstrated increased sensitivity for platelet P-selectin expression and phosphatidylserine exposure in response to platelet GPVI or PAR agonists compared with lean NHPs. Obese NHPs exhibited elevated levels of C-reactive protein, cathepsin D, and myeloperoxidase compared with lean NHPs. Following pharmacological inhibition of FIX activation by FXIa, platelet priming for activation by GPVI or PAR agonists, C-reactive protein levels, and endothelial VCAM-1 levels were reduced in obese NHPs. Conclusion: FXI activation promotes the proinflammatory phenotype of hyperlipidemia by priming platelet activation and inciting endothelial cell dysfunction.

20.
Blood ; 118(25): 6481-2, 2011 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-22174307

RESUMEN

In this issue of Blood, Yau and colleagues provide evidence that guide catheter thrombosis in patients undergoing percutaneous coronary intervention (PCI) is initiated by contact system activation, and that unfractionated heparin is more effective than fondaparinux at limiting catheter occlusions because heparin-antithrombin complexes are able to target multiple points along the coagulation cascade, as opposed to the smaller fractionated heparins and heparinoids that provide higher selectivity for factor Xa.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA